1
|
Duong TB, Fernandes AT, Ravisankar P, Talbot JC, Waxman JS. Tbx1-dependent and independent pathways promote six gene expression downstream of retinoic acid signaling to determine cardiomyocyte number in zebrafish. Dev Biol 2025; 524:17-28. [PMID: 40311730 PMCID: PMC12146055 DOI: 10.1016/j.ydbio.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 03/26/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Tight regulation of retinoic acid (RA) levels is critical for normal heart development in vertebrates, with early RA signaling restricting the size of the cardiac progenitor field within the anterior lateral plate mesoderm (ALPM). However, the regulatory networks by which RA signaling limits the size of the cardiac progenitor field and consequently cardiomyocyte (CM) number are not fully understood. Here, we identified that the expression of the transcription factors six1b and six2a, whose orthologs regulate outflow tract (OFT) development in mice, are expanded within the ALPM of RA-deficient zebrafish embryos. At 48 h post-fertilization (hpf), RA-deficient six1b;six2a double mutants, but not single six1b or six2a mutants, had a reduction in the number of surplus CMs relative to RA-deficient wild-type embryos. The expansion of six1b, as well as fgf8a, within the ALPM were dependent on tbx1, a factor that is also expanded within the ALPM of RA-deficient zebrafish embryos. However, the restriction of six2a expression by RA was independent of Tbx1. Consistent with a bifurcation of pathways downstream of RA signaling, loss of function experiments demonstrates that tbx1 expansion alone does not contribute to the surplus CMs in RA-deficient embryos. Together, our data indicate that both Tbx1-dependent and independent pathways restrict Six dosage downstream of RA within the ALPM to pattern the CM progenitor field and establish proper heart size.
Collapse
Affiliation(s)
- Tiffany B Duong
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew T Fernandes
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Padmapriyadarshini Ravisankar
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jared C Talbot
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
2
|
Griffin AHC, Small AM, Johnson RD, Medina AM, Kollar KT, Nazir RA, McGuire AM, Schumacher JA. Retinoic acid promotes second heart field addition and regulates ventral aorta patterning in zebrafish. Dev Biol 2025; 522:143-155. [PMID: 40147741 DOI: 10.1016/j.ydbio.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 03/09/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Retinoic acid (RA) signaling is used reiteratively during vertebrate heart development. Its earliest known role is to restrict formation of the earlier-differentiating first heart field (FHF) progenitors, while promoting the differentiation of second heart field (SHF) progenitors that give rise to the arterial pole of the ventricle and outflow tract (OFT). However, requirements for RA signaling at later stages of cardiogenesis remain poorly understood. Here, we investigated the role of RA signaling after the later differentiating SHF cells have begun to add to the OFT. We found that inhibiting RA production in zebrafish beginning at 26 hours post fertilization (hpf) produced embryos that have smaller ventricles with fewer ventricular cardiomyocytes, and reduced number of smooth muscle cells in the bulbus arteriosus (BA) of the OFT. Our results suggest that the deficiency of the ventricular cardiomyocytes is due to reduced SHF addition to the arterial pole. In contrast to smaller ventricles and BA, later RA deficiency also results in a dramatically elongated posterior branch of the adjacent ventral aorta, which is surrounded by an increased number of smooth muscle cells. Altogether, our results reveal that RA signaling is required during the period of SHF addition to promote addition of ventricular cardiomyocytes, partition smooth muscle cells onto the BA and posterior ventral aorta, and to establish proper ventral aorta anterior-posterior patterning.
Collapse
Affiliation(s)
| | - Allison M Small
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Riley D Johnson
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Anna M Medina
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Kiki T Kollar
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Ridha A Nazir
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | | | - Jennifer A Schumacher
- Department of Biology, Miami University, Oxford, OH, 45056, USA; Department of Biological Sciences, Miami University, Hamilton, OH, 45011, USA.
| |
Collapse
|
3
|
Angom RS, Singh M, Muhammad H, Varanasi SM, Mukhopadhyay D. Zebrafish as a Versatile Model for Cardiovascular Research: Peering into the Heart of the Matter. Cells 2025; 14:531. [PMID: 40214485 PMCID: PMC11988917 DOI: 10.3390/cells14070531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the world. A total of 17.5 million people died of CVDs in the year 2012, accounting for 31% of all deaths globally. Vertebrate animal models have been used to understand cardiac disease biology, as the cellular, molecular, and physiological aspects of human CVDs can be replicated closely in these organisms. Zebrafish is a popular model organism offering an arsenal of genetic tools that allow the rapid in vivo analysis of vertebrate gene function and disease conditions. It has a short breeding cycle, high fecundity, optically transparent embryos, rapid internal organ development, and easy maintenance. This review aims to give readers an overview of zebrafish cardiac biology and a detailed account of heart development in zebrafish and its comparison with humans and the conserved genetic circuitry. We also discuss the contributions made in CVD research using the zebrafish model. The first part of this review focuses on detailed information on the morphogenetic and differentiation processes in early cardiac development. The overlap and divergence of the human heart's genetic circuitry, structure, and physiology are emphasized wherever applicable. In the second part of the review, we overview the molecular tools and techniques available to dissect gene function and expression in zebrafish, with special mention of the use of these tools in cardiac biology.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Meghna Singh
- Department of Pathology and Lab Medicine, University of California, Los Angeles, CA 92093, USA;
| | - Huzaifa Muhammad
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Sai Manasa Varanasi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| |
Collapse
|
4
|
Montero-Herradón S, García-Ceca J, Zapata AG. Thymus Ontogeny and Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:21-49. [PMID: 40067583 DOI: 10.1007/978-3-031-77921-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The thymus is a primary lymphoid organ composed of a three-dimensional (3D) epithelial network that provides a specialized microenvironment for the phenotypical and functional maturation of lymphoid progenitors. The specification of the pharyngeal endoderm to thymus fate occurs during the early stages of thymic organogenesis, independent of the expression of the transcription factor Foxn1. However, Foxn1 governs the later organogenesis of thymus together with the colonizing lymphoid cells. In the present chapter, we will review recent evidence on the topic covered in our original chapter (Muñoz and Zapata 2019). It described the early development of thymus and its resemblance to the development of endoderm-derived epithelial organs based on tubulogenesis and branching morphogenesis as well as the molecules known to be involved in these processes.
Collapse
Affiliation(s)
- Sara Montero-Herradón
- Department of Cell Biology. Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain
- Health Research Institute, Madrid, Spain
| | - Javier García-Ceca
- Department of Cell Biology. Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain
- Health Research Institute, Madrid, Spain
| | - Agustín G Zapata
- Department of Cell Biology. Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain.
- Health Research Institute, Madrid, Spain.
| |
Collapse
|
5
|
Fainsod A, Vadigepalli R. Rethinking retinoic acid self-regulation: A signaling robustness network approach. Curr Top Dev Biol 2024; 161:113-141. [PMID: 39870431 DOI: 10.1016/bs.ctdb.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
All-trans retinoic acid (ATRA) signaling is a major pathway regulating numerous differentiation, proliferation, and patterning processes throughout life. ATRA biosynthesis depends on the nutritional availability of vitamin A and other retinoids and carotenoids, while it is sensitive to dietary and environmental toxicants. This nutritional and environmental influence requires a robustness response that constantly fine-tunes the ATRA metabolism to maintain a context-specific, physiological range of signaling levels. The ATRA metabolic and signaling network is characterized by the existence of multiple enzymes, transcription factors, and binding proteins capable of performing the same activity. The partial spatiotemporal expression overlap of these enzymes and proteins yields different network compositions in the cells and tissues where this pathway is active. Genetic polymorphisms affecting the activity of individual network components further impact the network composition variability and the self-regulatory feedback response to ATRA fluctuations. Experiments directly challenging the robustness response uncovered a Pareto optimality in the ATRA network, such that some genetic backgrounds efficiently deal with excess ATRA but are very limited in their robustness response to reduced ATRA and vice versa. We discuss a network-focused framework to describe the robustness response and the Pareto optimality of the ATRA metabolic and signaling network.
Collapse
Affiliation(s)
- Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology and Genomic Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
6
|
Duester G. Early retinoic acid signaling organizes the body axis and defines domains for the forelimb and eye. Curr Top Dev Biol 2024; 161:1-32. [PMID: 39870430 PMCID: PMC11969570 DOI: 10.1016/bs.ctdb.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
All-trans RA (ATRA) is a small molecule derived from retinol (vitamin A) that directly controls gene expression at the transcriptional level by serving as a ligand for nuclear ATRA receptors. ATRA is produced by ATRA-generating enzymes that convert retinol to retinaldehyde (retinol dehydrogenase; RDH10) followed by conversion of retinaldehyde to ATRA (retinaldehyde dehydrogenase; ALDH1A1, ALDH1A2, or ALDH1A3). Determining what ATRA normally does during vertebrate development has been challenging as studies employing ATRA gain-of-function (RA treatment) often do not agree with genetic loss-of-function studies that remove ATRA via knockouts of ATRA-generating enzymes. In mouse embryos, ATRA is first generated at stage E7.5 by ATRA-generating enzymes whose genes are first expressed at that stage. This article focuses upon what ATRA normally does at early stages based upon these knockout studies. It has been observed that early-generated ATRA performs three essential functions: (1) activation of genes that control hindbrain and spinal cord patterning; (2) repression of Fgf8 in the heart field and caudal progenitors to provide an FGF8-free region in the trunk essential for somitogenesis, heart morphogenesis, and initiation of forelimb fields; and (3) actions that stimulate invagination of the optic vesicle to form the optic cup.
Collapse
Affiliation(s)
- Gregg Duester
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States.
| |
Collapse
|
7
|
Dumas CE, Rousset C, De Bono C, Cortés C, Jullian E, Lescroart F, Zaffran S, Adachi N, Kelly RG. Retinoic acid signalling regulates branchiomeric neck muscle development at the head/trunk interface. Development 2024; 151:dev202905. [PMID: 39082789 DOI: 10.1242/dev.202905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/18/2024] [Indexed: 08/30/2024]
Abstract
Skeletal muscles of the head and trunk originate in distinct lineages with divergent regulatory programmes converging on activation of myogenic determination factors. Branchiomeric head and neck muscles share a common origin with cardiac progenitor cells in cardiopharyngeal mesoderm (CPM). The retinoic acid (RA) signalling pathway is required during a defined early time window for normal deployment of cells from posterior CPM to the heart. Here, we show that blocking RA signalling in the early mouse embryo also results in selective loss of the trapezius neck muscle, without affecting other skeletal muscles. RA signalling is required for robust expression of myogenic determination factors in posterior CPM and subsequent expansion of the trapezius primordium. Lineage-specific activation of a dominant-negative RA receptor reveals that trapezius development is not regulated by direct RA signalling to myogenic progenitor cells in CPM, or through neural crest cells, but indirectly through the somitic lineage, closely apposed with posterior CPM in the early embryo. These findings suggest that trapezius development is dependent on precise spatiotemporal interactions between cranial and somitic mesoderm at the head/trunk interface.
Collapse
Affiliation(s)
- Camille E Dumas
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Célia Rousset
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | | | - Claudio Cortés
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Estelle Jullian
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | | | - Stéphane Zaffran
- Aix-Marseille Université, INSERM, MMG U1251, 13005 Marseille, France
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| |
Collapse
|
8
|
Bayramov AV, Yastrebov SA, Mednikov DN, Araslanova KR, Ermakova GV, Zaraisky AG. Paired fins in vertebrate evolution and ontogeny. Evol Dev 2024; 26:e12478. [PMID: 38650470 DOI: 10.1111/ede.12478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 02/28/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
The origin of paired appendages became one of the most important adaptations of vertebrates, allowing them to lead active lifestyles and explore a wide range of ecological niches. The basic form of paired appendages in evolution is the fins of fishes. The problem of paired appendages has attracted the attention of researchers for more than 150 years. During this time, a number of theories have been proposed, mainly based on morphological data, two of which, the Balfour-Thacher-Mivart lateral fold theory and Gegenbaur's gill arch theory, have not lost their relevance. So far, however, none of the proposed ideas has been supported by decisive evidence. The study of the evolutionary history of the appearance and development of paired appendages lies at the intersection of several disciplines and involves the synthesis of paleontological, morphological, embryological, and genetic data. In this review, we attempt to summarize and discuss the results accumulated in these fields and to analyze the theories put forward regarding the prerequisites and mechanisms that gave rise to paired fins and limbs in vertebrates.
Collapse
Affiliation(s)
- Andrey V Bayramov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Sergey A Yastrebov
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry N Mednikov
- Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | - Karina R Araslanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Galina V Ermakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Andrey G Zaraisky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Department of Regenerative Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
9
|
Moreno-Oñate M, Gallardo-Fuentes L, Martínez-García PM, Naranjo S, Jiménez-Gancedo S, Tena JJ, Santos-Pereira JM. Rewiring of the epigenome and chromatin architecture by exogenously induced retinoic acid signaling during zebrafish embryonic development. Nucleic Acids Res 2024; 52:3682-3701. [PMID: 38321954 PMCID: PMC11040003 DOI: 10.1093/nar/gkae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 02/08/2024] Open
Abstract
Retinoic acid (RA) is the ligand of RA receptors (RARs), transcription factors that bind to RA response elements. RA signaling is required for multiple processes during embryonic development, including body axis extension, hindbrain antero-posterior patterning and forelimb bud initiation. Although some RA target genes have been identified, little is known about the genome-wide effects of RA signaling during in vivo embryonic development. Here, we stimulate the RA pathway by treating zebrafish embryos with all-trans-RA (atRA) and use a combination of RNA-seq, ATAC-seq, ChIP-seq and HiChIP to gain insight into the molecular mechanisms by which exogenously induced RA signaling controls gene expression. We find that RA signaling is involved in anterior/posterior patterning, central nervous system development, and the transition from pluripotency to differentiation. AtRA treatment also alters chromatin accessibility during early development and promotes chromatin binding of RARαa and the RA targets Hoxb1b, Meis2b and Sox3, which cooperate in central nervous system development. Finally, we show that exogenous RA induces a rewiring of chromatin architecture, with alterations in chromatin 3D interactions involving target genes. Altogether, our findings identify genome-wide targets of RA signaling and provide a molecular mechanism by which developmental signaling pathways regulate target gene expression by altering chromatin topology.
Collapse
Affiliation(s)
- Marta Moreno-Oñate
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Lourdes Gallardo-Fuentes
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Pedro M Martínez-García
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Silvia Naranjo
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Sandra Jiménez-Gancedo
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Juan J Tena
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - José M Santos-Pereira
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Biología Celular, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| |
Collapse
|
10
|
Yuikawa T, Sato T, Ikeda M, Tsuruoka M, Yasuda K, Sato Y, Nasu K, Yamasu K. Elongation of the developing spinal cord is driven by Oct4-type transcription factor-mediated regulation of retinoic acid signaling in zebrafish embryos. Dev Dyn 2024; 253:404-422. [PMID: 37850839 DOI: 10.1002/dvdy.666] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Elongation of the spinal cord is dependent on neural development from neuromesodermal progenitors in the tail bud. We previously showed the involvement of the Oct4-type gene, pou5f3, in this process in zebrafish mainly by dominant-interference gene induction, but, to compensate for the limitation of this transgene approach, mutant analysis was indispensable. pou5f3 involvement in the signaling pathways was another unsolved question. RESULTS We examined the phenotypes of pou5f3 mutants and the effects of Pou5f3 activation by the tamoxifen-ERT2 system in the posterior neural tube, together confirming the involvement of pou5f3. The reporter assays using P19 cells implicated tail bud-related transcription factors in pou5f3 expression. Regulation of tail bud development by retinoic acid (RA) signaling was confirmed by treatment of embryos with RA and the synthesis inhibitor, and in vitro reporter assays further showed that RA signaling regulated pou5f3 expression. Importantly, the expression of the RA degradation enzyme gene, cyp26a1, was down-regulated in embryos with disrupted pou5f3 activity. CONCLUSIONS The involvement of pou5f3 in spinal cord extension was supported by using mutants and the gain-of-function approach. Our findings further suggest that pou5f3 regulates the RA level, contributing to neurogenesis in the posterior neural tube.
Collapse
Affiliation(s)
- Tatsuya Yuikawa
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Takehisa Sato
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Masaaki Ikeda
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Momo Tsuruoka
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Kaede Yasuda
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Yuto Sato
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Kouhei Nasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama City, Saitama, Japan
| |
Collapse
|
11
|
Martins-Costa C, Wilson V, Binagui-Casas A. Neuromesodermal specification during head-to-tail body axis formation. Curr Top Dev Biol 2024; 159:232-271. [PMID: 38729677 DOI: 10.1016/bs.ctdb.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The anterior-to-posterior (head-to-tail) body axis is extraordinarily diverse among vertebrates but conserved within species. Body axis development requires a population of axial progenitors that resides at the posterior of the embryo to sustain elongation and is then eliminated once axis extension is complete. These progenitors occupy distinct domains in the posterior (tail-end) of the embryo and contribute to various lineages along the body axis. The subset of axial progenitors with neuromesodermal competency will generate both the neural tube (the precursor of the spinal cord), and the trunk and tail somites (producing the musculoskeleton) during embryo development. These axial progenitors are called Neuromesodermal Competent cells (NMCs) and Neuromesodermal Progenitors (NMPs). NMCs/NMPs have recently attracted interest beyond the field of developmental biology due to their clinical potential. In the mouse, the maintenance of neuromesodermal competency relies on a fine balance between a trio of known signals: Wnt/β-catenin, FGF signalling activity and suppression of retinoic acid signalling. These signals regulate the relative expression levels of the mesodermal transcription factor Brachyury and the neural transcription factor Sox2, permitting the maintenance of progenitor identity when co-expressed, and either mesoderm or neural lineage commitment when the balance is tilted towards either Brachyury or Sox2, respectively. Despite important advances in understanding key genes and cellular behaviours involved in these fate decisions, how the balance between mesodermal and neural fates is achieved remains largely unknown. In this chapter, we provide an overview of signalling and gene regulatory networks in NMCs/NMPs. We discuss mutant phenotypes associated with axial defects, hinting at the potential significant role of lesser studied proteins in the maintenance and differentiation of the progenitors that fuel axial elongation.
Collapse
Affiliation(s)
- C Martins-Costa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - V Wilson
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| | - A Binagui-Casas
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
12
|
Bedois AMH, Parker HJ, Price AJ, Morrison JA, Bronner ME, Krumlauf R. Sea lamprey enlightens the origin of the coupling of retinoic acid signaling to vertebrate hindbrain segmentation. Nat Commun 2024; 15:1538. [PMID: 38378737 PMCID: PMC10879103 DOI: 10.1038/s41467-024-45911-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 02/06/2024] [Indexed: 02/22/2024] Open
Abstract
Retinoic acid (RA) is involved in antero-posterior patterning of the chordate body axis and, in jawed vertebrates, has been shown to play a major role at multiple levels of the gene regulatory network (GRN) regulating hindbrain segmentation. Knowing when and how RA became coupled to the core hindbrain GRN is important for understanding how ancient signaling pathways and patterning genes can evolve and generate diversity. Hence, we investigated the link between RA signaling and hindbrain segmentation in the sea lamprey Petromyzon marinus, an important jawless vertebrate model providing clues to decipher ancestral vertebrate features. Combining genomics, gene expression, and functional analyses of major components involved in RA synthesis (Aldh1as) and degradation (Cyp26s), we demonstrate that RA signaling is coupled to hindbrain segmentation in lamprey. Thus, the link between RA signaling and hindbrain segmentation is a pan vertebrate feature of the hindbrain and likely evolved at the base of vertebrates.
Collapse
Affiliation(s)
- Alice M H Bedois
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Hugo J Parker
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Andrew J Price
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Jason A Morrison
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Robb Krumlauf
- Stowers Institute for Medical Research, Kansas City, MO, 64110, USA.
- Department of Anatomy and Cell Biology, Kansas University Medical Center, Kansas City, MO, 66160, USA.
| |
Collapse
|
13
|
Bedois AMH, Parker HJ, Bronner ME, Krumlauf R. Sea lamprey enlightens the origin of the coupling of retinoic acid signaling to vertebrate hindbrain segmentation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.07.548143. [PMID: 37461675 PMCID: PMC10350081 DOI: 10.1101/2023.07.07.548143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Retinoic acid (RA) is involved in antero-posterior patterning of the chordate body axis and, in jawed vertebrates, has been shown to play a major role at multiple levels of the gene regulatory network (GRN) regulating hindbrain segmentation. Knowing when and how RA became coupled to the core hindbrain GRN is important for understanding how ancient signaling pathways and patterning genes can evolve and generate diversity. Hence, we investigated the link between RA signaling and hindbrain segmentation in the sea lamprey Petromyzon marinus, an important jawless vertebrate model providing clues to decipher ancestral vertebrate features. Combining genomics, gene expression, and functional analyses of major components involved in RA synthesis (Aldh1as) and degradation (Cyp26s), we demonstrate that RA signaling is coupled to hindbrain segmentation in lamprey. Thus, the link between RA signaling and hindbrain segmentation is a pan vertebrate feature of the hindbrain and likely evolved at the base of vertebrates.
Collapse
Affiliation(s)
- Alice M. H. Bedois
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
| | - Hugo J. Parker
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
| | - Marianne E. Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Robb Krumlauf
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA
- Department of Anatomy and Cell Biology, Kansas University Medical Center, Kansas City, Kansas 66160, USA
| |
Collapse
|
14
|
Čapek D, Safroshkin M, Morales-Navarrete H, Toulany N, Arutyunov G, Kurzbach A, Bihler J, Hagauer J, Kick S, Jones F, Jordan B, Müller P. EmbryoNet: using deep learning to link embryonic phenotypes to signaling pathways. Nat Methods 2023; 20:815-823. [PMID: 37156842 PMCID: PMC10250202 DOI: 10.1038/s41592-023-01873-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 04/05/2023] [Indexed: 05/10/2023]
Abstract
Evolutionarily conserved signaling pathways are essential for early embryogenesis, and reducing or abolishing their activity leads to characteristic developmental defects. Classification of phenotypic defects can identify the underlying signaling mechanisms, but this requires expert knowledge and the classification schemes have not been standardized. Here we use a machine learning approach for automated phenotyping to train a deep convolutional neural network, EmbryoNet, to accurately identify zebrafish signaling mutants in an unbiased manner. Combined with a model of time-dependent developmental trajectories, this approach identifies and classifies with high precision phenotypic defects caused by loss of function of the seven major signaling pathways relevant for vertebrate development. Our classification algorithms have wide applications in developmental biology and robustly identify signaling defects in evolutionarily distant species. Furthermore, using automated phenotyping in high-throughput drug screens, we show that EmbryoNet can resolve the mechanism of action of pharmaceutical substances. As part of this work, we freely provide more than 2 million images that were used to train and test EmbryoNet.
Collapse
Affiliation(s)
- Daniel Čapek
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | | | - Hernán Morales-Navarrete
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
- Centre for the Advanced Study of Collective Behaviour, Konstanz, Germany
| | - Nikan Toulany
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | | | - Anica Kurzbach
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
| | - Johanna Bihler
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Julia Hagauer
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Sebastian Kick
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Felicity Jones
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| | - Ben Jordan
- Systems Biology of Development, University of Konstanz, Konstanz, Germany
| | - Patrick Müller
- Systems Biology of Development, University of Konstanz, Konstanz, Germany.
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany.
- Centre for the Advanced Study of Collective Behaviour, Konstanz, Germany.
| |
Collapse
|
15
|
Hawkins MR, Wingert RA. Zebrafish as a Model to Study Retinoic Acid Signaling in Development and Disease. Biomedicines 2023; 11:biomedicines11041180. [PMID: 37189798 DOI: 10.3390/biomedicines11041180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Retinoic acid (RA) is a metabolite of vitamin A (retinol) that plays various roles in development to influence differentiation, patterning, and organogenesis. RA also serves as a crucial homeostatic regulator in adult tissues. The role of RA and its associated pathways are well conserved from zebrafish to humans in both development and disease. This makes the zebrafish a natural model for further interrogation into the functions of RA and RA-associated maladies for the sake of basic research, as well as human health. In this review, we explore both foundational and recent studies using zebrafish as a translational model for investigating RA from the molecular to the organismal scale.
Collapse
Affiliation(s)
- Matthew R Hawkins
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
16
|
Udaykumar N, Zaidi MAA, Rai A, Sen J. CNKSR2, a downstream mediator of retinoic acid signaling, modulates the Ras/Raf/MEK pathway to regulate patterning and invagination of the chick forebrain roof plate. Development 2023; 150:286897. [PMID: 36734326 DOI: 10.1242/dev.200857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 01/03/2023] [Indexed: 02/04/2023]
Abstract
During embryonic development, the forebrain roof plate undergoes invagination, leading to separation of the cerebral hemispheres. Any defects in this process, in humans, lead to middle interhemispheric holoprosencephaly (MIH-HPE). In this study, we have identified a previously unreported downstream mediator of retinoic acid (RA) signaling, CNKSR2, which is expressed in the forebrain roof plate in the chick embryo. Knockdown of CNKSR2 affects invagination, cell proliferation and patterning of the roof plate, similar to the phenotypes observed upon inhibition of RA signaling. We further demonstrate that CNKSR2 functions by modulating the Ras/Raf/MEK signaling. This appears to be crucial for patterning of the forebrain roof plate and its subsequent invagination, leading to the formation of the cerebral hemispheres. Thus, a set of novel molecular players have been identified that regulate the morphogenesis of the avian forebrain.
Collapse
Affiliation(s)
- Niveda Udaykumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016 Uttar Pradesh, India
| | - Mohd Ali Abbas Zaidi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016 Uttar Pradesh, India
| | - Aishwarya Rai
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016 Uttar Pradesh, India
| | - Jonaki Sen
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016 Uttar Pradesh, India
- Mehta Family Center for Engineering in Medicine (MFCEM), Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| |
Collapse
|
17
|
Abstract
Vitamin A (retinol) is an important nutrient for embryonic development and adult health. Early studies identified retinoic acid (RA) as a metabolite of retinol, however, its importance was not apparent. Later, it was observed that RA treatment of vertebrate embryos had teratogenic effects on limb development. Subsequently, the discovery of nuclear RA receptors (RARs) revealed that RA controls gene expression directly at the transcriptional level through a process referred to as RA signaling. This important discovery led to further studies demonstrating that RA and RARs are required for normal embryonic development. The determination of RA function during normal development has been challenging as RA gain-of-function studies often lead to conclusions about normal development that conflict with RAR or RA loss-of-function studies. However, genetic loss-of-function studies have identified direct target genes of endogenous RA/RAR that are required for normal development of specific tissues. Thus, genetic loss-of-function studies that eliminate RARs or RA-generating enzymes have been instrumental in revealing that RA signaling is required for normal early development of many organs and tissues, including the hindbrain, posterior body axis, somites, spinal cord, forelimbs, heart, and eye.
Collapse
Affiliation(s)
- Marie Berenguer
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Gregg Duester
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
18
|
Martin BL, Steventon B. A fishy tail: Insights into the cell and molecular biology of neuromesodermal cells from zebrafish embryos. Dev Biol 2022; 487:67-73. [PMID: 35525020 DOI: 10.1016/j.ydbio.2022.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/29/2022] [Accepted: 04/26/2022] [Indexed: 11/03/2022]
Abstract
Vertebrate embryos establish their primary body axis in a conserved progressive fashion from the anterior to the posterior. During this process, a posteriorly localized neuromesodermal cell population called neuromesodermal progenitors (NMps) plays a critical role in contributing new cells to the spinal cord and mesoderm as the embryo elongates. Defects in neuromesodermal population development can cause severe disruptions to the formation of the body posterior to the head. Given their importance during development and their potential, some of which has already been realized, for revealing new methods of in vitro tissue generation, there is great interest in better understanding NMp biology. The zebrafish model system has been instrumental in advancing our understanding of the molecular and cellular attributes of the NM cell population and its derivatives. In this review, we focus on our current understanding of the zebrafish NM population and its contribution to body axis formation, with particular emphasis on the lineage potency, morphogenesis, and niche factors that promote or inhibit differentiation.
Collapse
Affiliation(s)
- Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794-5215, USA.
| | - Benjamin Steventon
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
19
|
Mackowetzky K, Dicipulo R, Fox SC, Philibert DA, Todesco H, Doshi JD, Kawakami K, Tierney K, Waskiewicz AJ. Retinoic acid signaling regulates late stages of semicircular canal morphogenesis and otolith maintenance in the zebrafish inner ear. Dev Dyn 2022; 251:1798-1815. [PMID: 35710880 DOI: 10.1002/dvdy.510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The vitamin A derivative all-trans retinoic acid (RA) regulates early stages of inner ear development. As the early disruption of the RA pathway results in profound mispatterning of the developing inner ear, this confounds analyses of specific roles in later stages. Therefore, we used the temporal-specific exposure of all-trans RA or diethylaminobenzaldehyde to evaluate RA functions in late otic development. RESULTS Perturbing late RA signaling causes behavioral defects analogous to those expected in larvae suffering from vestibular dysfunction. These larvae also demonstrate malformations of the semi-circular canals, as visualized through (a) use of the transgenic strain nkhspdmc12a, a fluorescent reporter expressed in otic epithelium; and (b) injection of the fluorescent lipophilic dye DiI. We also noted the altered expression of genes encoding ECM proteins or modifying enzymes. Other malformations of the inner ear observed in our work include the loss or reduced size of the utricular and saccular otoliths, suggesting a role for RA in otolith maintenance. CONCLUSION Our work has identified a previously undescribed late phase of RA activity in otic development, demonstrating that vestibular defects observed in human patients in relation to perturbed RA signaling are not solely due to its early disruption in otic development.
Collapse
Affiliation(s)
- Kacey Mackowetzky
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Renée Dicipulo
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Sabrina C Fox
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,Women & Children's Health Research Institute, Edmonton, Alberta, Canada
| | - Danielle A Philibert
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Hayley Todesco
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Jainil D Doshi
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, Shizuoka, Japan
| | - Keith Tierney
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew J Waskiewicz
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,Women & Children's Health Research Institute, Edmonton, Alberta, Canada
| |
Collapse
|
20
|
Gur M, Edri T, Moody SA, Fainsod A. Retinoic Acid is Required for Normal Morphogenetic Movements During Gastrulation. Front Cell Dev Biol 2022; 10:857230. [PMID: 35531100 PMCID: PMC9068879 DOI: 10.3389/fcell.2022.857230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/21/2022] [Indexed: 11/23/2022] Open
Abstract
Retinoic acid (RA) is a central regulatory signal that controls numerous developmental processes in vertebrate embryos. Although activation of Hox expression is considered one of the earliest functions of RA signaling in the embryo, there is evidence that embryos are poised to initiate RA signaling just before gastrulation begins, and manipulations of the RA pathway have been reported to show gastrulation defects. However, which aspects of gastrulation are affected have not been explored in detail. We previously showed that partial inhibition of RA biosynthesis causes a delay in the rostral migration of some of the earliest involuting cells, the leading edge mesendoderm (LEM) and the prechordal mesoderm (PCM). Here we identify several detrimental gastrulation defects resulting from inhibiting RA biosynthesis by three different treatments. RA reduction causes a delay in the progression through gastrulation as well as the rostral migration of the goosecoid-positive PCM cells. RA inhibition also hampered the elongation of explanted dorsal marginal zones, the compaction of the blastocoel, and the length of Brachet’s cleft, all of which indicate an effect on LEM/PCM migration. The cellular mechanisms underlying this deficit were shown to include a reduced deposition of fibronectin along Brachet’s cleft, the substrate for their migration, as well as impaired separation of the blastocoel roof and involuting mesoderm, which is important for the formation of Brachet’s cleft and successful LEM/PCM migration. We further show reduced non-canonical Wnt signaling activity and altered expression of genes in the Ephrin and PDGF signaling pathways, both of which are required for the rostral migration of the LEM/PCM, following RA reduction. Together, these experiments demonstrate that RA signaling performs a very early function critical for the progression of gastrulation morphogenetic movements.
Collapse
Affiliation(s)
- Michal Gur
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tamir Edri
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sally A. Moody
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, Washington, DC, United States
- *Correspondence: Sally A. Moody, ; Abraham Fainsod,
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- *Correspondence: Sally A. Moody, ; Abraham Fainsod,
| |
Collapse
|
21
|
Gur M, Bendelac-Kapon L, Shabtai Y, Pillemer G, Fainsod A. Reduced Retinoic Acid Signaling During Gastrulation Induces Developmental Microcephaly. Front Cell Dev Biol 2022; 10:844619. [PMID: 35372345 PMCID: PMC8967241 DOI: 10.3389/fcell.2022.844619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/24/2022] [Indexed: 12/21/2022] Open
Abstract
Retinoic acid (RA) is a central signaling molecule regulating multiple developmental decisions during embryogenesis. Excess RA induces head malformations, primarily by expansion of posterior brain structures at the expense of anterior head regions, i.e., hindbrain expansion. Despite this extensively studied RA teratogenic effect, a number of syndromes exhibiting microcephaly, such as DiGeorge, Vitamin A Deficiency, Fetal Alcohol Syndrome, and others, have been attributed to reduced RA signaling. This causative link suggests a requirement for RA signaling during normal head development in all these syndromes. To characterize this novel RA function, we studied the involvement of RA in the early events leading to head formation in Xenopus embryos. This effect was mapped to the earliest RA biosynthesis in the embryo within the gastrula Spemann-Mangold organizer. Head malformations were observed when reduced RA signaling was induced in the endogenous Spemann-Mangold organizer and in the ectopic organizer of twinned embryos. Two embryonic retinaldehyde dehydrogenases, ALDH1A2 (RALDH2) and ALDH1A3 (RALDH3) are initially expressed in the organizer and subsequently mark the trunk and the migrating leading edge mesendoderm, respectively. Gene-specific knockdowns and CRISPR/Cas9 targeting show that RALDH3 is a key enzyme involved in RA production required for head formation. These observations indicate that in addition to the teratogenic effect of excess RA on head development, RA signaling also has a positive and required regulatory role in the early formation of the head during gastrula stages. These results identify a novel RA activity that concurs with its proposed reduction in syndromes exhibiting microcephaly.
Collapse
|
22
|
Paulissen E, Palmisano NJ, Waxman J, Martin BL. Somite morphogenesis is required for axial blood vessel formation during zebrafish embryogenesis. eLife 2022; 11:74821. [PMID: 35137687 PMCID: PMC8863375 DOI: 10.7554/elife.74821] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Angioblasts that form the major axial blood vessels of the dorsal aorta and cardinal vein migrate toward the embryonic midline from distant lateral positions. Little is known about what controls the precise timing of angioblast migration and their final destination at the midline. Using zebrafish, we found that midline angioblast migration requires neighboring tissue rearrangements generated by somite morphogenesis. The somitic shape changes cause the adjacent notochord to separate from the underlying endoderm, creating a ventral midline cavity that provides a physical space for the angioblasts to migrate into. The anterior to posterior progression of midline angioblast migration is facilitated by retinoic acid-induced anterior to posterior somite maturation and the subsequent progressive opening of the ventral midline cavity. Our work demonstrates a critical role for somite morphogenesis in organizing surrounding tissues to facilitate notochord positioning and angioblast migration, which is ultimately responsible for creating a functional cardiovascular system.
Collapse
Affiliation(s)
- Eric Paulissen
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Nicholas J Palmisano
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| | - Joshua Waxman
- Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Benjamin Louis Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, United States
| |
Collapse
|
23
|
Enhanced Loss of Retinoic Acid Network Genes in Xenopus laevis Achieves a Tighter Signal Regulation. Cells 2022; 11:cells11030327. [PMID: 35159137 PMCID: PMC8834563 DOI: 10.3390/cells11030327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/10/2022] Open
Abstract
Retinoic acid (RA) is a major regulatory signal during embryogenesis produced from vitamin A (retinol) by an extensive, autoregulating metabolic and signaling network to prevent fluctuations that result in developmental malformations. Xenopus laevis is an allotetraploid hybrid frog species whose genome includes L (long) and S (short) chromosomes from the originating species. Evolutionarily, the X. laevis subgenomes have been losing either L or S homoeologs in about 43% of genes to generate singletons. In the RA network, out of the 47 genes, about 47% have lost one of the homoeologs, like the genome average. Interestingly, RA metabolism genes from storage (retinyl esters) to retinaldehyde production exhibit enhanced gene loss with 75% singletons out of 28 genes. The effect of this gene loss on RA signaling autoregulation was studied. Employing transient RA manipulations, homoeolog gene pairs were identified in which one homoeolog exhibits enhanced responses or looser regulation than the other, while in other pairs both homoeologs exhibit similar RA responses. CRISPR/Cas9 targeting of individual homoeologs to reduce their activity supports the hypothesis where the RA metabolic network gene loss results in tighter network regulation and more efficient RA robustness responses to overcome complex regulation conditions.
Collapse
|
24
|
Dickinson AJG, Turner SD, Wahl S, Kennedy AE, Wyatt BH, Howton DA. E-liquids and vanillin flavoring disrupts retinoic acid signaling and causes craniofacial defects in Xenopus embryos. Dev Biol 2022; 481:14-29. [PMID: 34543654 PMCID: PMC8665092 DOI: 10.1016/j.ydbio.2021.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 01/03/2023]
Abstract
Environmental teratogens such as smoking are known risk factors for developmental disorders such as cleft palate. While smoking rates have declined, a new type of smoking, called vaping is on the rise. Vaping is the use of e-cigarettes to vaporize and inhale an e-liquid containing nicotine and food-like flavors. There is the potential that, like smoking, vaping could also pose a danger to the developing human. Rather than waiting for epidemiological and mammalian studies, we have turned to an aquatic developmental model, Xenopus laevis, to more quickly assess whether e-liquids contain teratogens that could lead to craniofacial malformations. Xenopus, like zebrafish, has the benefit of being a well-established developmental model and has also been effective in predicting whether a chemical could be a teratogen. We have determined that embryonic exposure to dessert flavored e-liquids can cause craniofacial abnormalities, including an orofacial cleft in Xenopus. To better understand the underlying mechanisms contributing to these defects, transcriptomic analysis of the facial tissues of embryos exposed to a representative dessert flavored e-liquid vapor extract was performed. Analysis of differentially expressed genes in these embryos revealed several genes associated with retinoic acid metabolism or the signaling pathway. Consistently, retinoic acid receptor inhibition phenocopied the craniofacial defects as those embryos exposed to the vapor extract of the e-liquid. Such malformations also correlated with a group of common differentially expressed genes, two of which are associated with midface birth defects in humans. Further, e-liquid exposure sensitized embryos to forming craniofacial malformations when they already had depressed retinoic acid signaling. Moreover, 13-cis-retinoic acid treatment could significantly reduce the e-liquid induced malformation in the midface. Such results suggest the possibility of an interaction between retinoic acid signaling and e-liquid exposure. One of the most popular and concentrated flavoring chemicals in dessert flavored e-liquids is vanillin. Xenopus embryos exposed to this chemical closely resembled embryos exposed to dessert-like e-liquids and a retinoic acid receptor antagonist. In summary, we determined that e-liquid chemicals, in particular vanillin, can cause craniofacial defects potentially by dysregulating retinoic acid signaling. This work warrants the evaluation of vanillin and other such flavoring additives in e-liquids on mammalian development.
Collapse
Affiliation(s)
| | - Stephen D Turner
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, USA; Signature Science LLC, Charlottesville, VA, USA
| | - Stacey Wahl
- Research and Education Department, Tompkins-McCaw Library for the Health Sciences, Virginia Commonwealth University, Richmond, VA, USA
| | - Allyson E Kennedy
- Directorate for Computer and Information Science and Engineering, National Science Foundation, Alexandria, VA, USA
| | - Brent H Wyatt
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, 27607, USA
| | - Deborah A Howton
- Department of Biology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
25
|
Duong TB, Waxman JS. Patterning of vertebrate cardiac progenitor fields by retinoic acid signaling. Genesis 2021; 59:e23458. [PMID: 34665508 DOI: 10.1002/dvg.23458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023]
Abstract
The influence of retinoic acid (RA) signaling on vertebrate development has a well-studied history. Cumulatively, we now understand that RA signaling has a conserved requirement early in development restricting cardiac progenitors within the anterior lateral plate mesoderm of vertebrate embryos. Moreover, genetic and pharmacological manipulations of RA signaling in vertebrate models have shown that proper heart development is achieved through the deployment of positive and negative feedback mechanisms, which maintain appropriate RA levels. In this brief review, we present a chronological overview of key work that has led to a current model of the critical role for early RA signaling in limiting the generation of cardiac progenitors within vertebrate embryos. Furthermore, we integrate the previous work in mice and our recent findings using zebrafish, which together show that RA signaling has remarkably conserved influences on the later-differentiating progenitor populations at the arterial and venous poles. We discuss how recognizing the significant conservation of RA signaling on the differentiation of these progenitor populations offers new perspectives and may impact future work dedicated to examining vertebrate heart development.
Collapse
Affiliation(s)
- Tiffany B Duong
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
26
|
Cho K, Lee SM, Heo J, Kwon YM, Chung D, Yu WJ, Bae SS, Choi G, Lee DS, Kim Y. Retinaldehyde Dehydrogenase Inhibition-Related Adverse Outcome Pathway: Potential Risk of Retinoic Acid Synthesis Inhibition during Embryogenesis. Toxins (Basel) 2021; 13:toxins13110739. [PMID: 34822523 PMCID: PMC8623920 DOI: 10.3390/toxins13110739] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 11/16/2022] Open
Abstract
Retinoic acid (RA) is one of the factors crucial for cell growth, differentiation, and embryogenesis; it interacts with the retinoic acid receptor and retinoic acid X receptor to eventually regulate target gene expression in chordates. RA is transformed from retinaldehyde via oxidization by retinaldehyde dehydrogenase (RALDH), which belongs to the family of oxidoreductases. Several chemicals, including disulphiram, diethylaminobenzaldehyde, and SB-210661, can effectively inhibit RALDH activity, potentially causing reproductive and developmental toxicity. The modes of action can be sequentially explained based on the molecular initiating event toward key events, and finally the adverse outcomes. Adverse outcome pathway (AOP) is a conceptual and theoretical framework that describes the sequential chain of casually liked events at different biological levels from molecular events to adverse effects. In the present review, we discussed a recently registered AOP (AOP297; inhibition of retinaldehyde dehydrogenase leads to population decline) to explain and support the weight of evidence for RALDH inhibition-related developmental toxicity using the existing knowledge.
Collapse
Affiliation(s)
- Kichul Cho
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Sang-Moo Lee
- Department of Applied Bioscience, Dong-A University, Busan 49315, Korea;
| | - Jina Heo
- Department of Growth Engine Research, Chungbuk Research Institute (CRI), Chungju 28517, Korea;
| | - Yong Min Kwon
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Dawoon Chung
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Woon-Jong Yu
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Seung Seob Bae
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Grace Choi
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Dae-Sung Lee
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
- Correspondence: (D.-S.L.); (Y.K.)
| | - Youngjun Kim
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Campus E 7.1, 66123 Saarbrücken, Germany
- Correspondence: (D.-S.L.); (Y.K.)
| |
Collapse
|
27
|
Parihar M, Bendelac-Kapon L, Gur M, Abbou T, Belorkar A, Achanta S, Kinberg K, Vadigepalli R, Fainsod A. Retinoic Acid Fluctuation Activates an Uneven, Direction-Dependent Network-Wide Robustness Response in Early Embryogenesis. Front Cell Dev Biol 2021; 9:747969. [PMID: 34746144 PMCID: PMC8564372 DOI: 10.3389/fcell.2021.747969] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/23/2021] [Indexed: 01/15/2023] Open
Abstract
Robustness is a feature of regulatory pathways to ensure signal consistency in light of environmental changes or genetic polymorphisms. The retinoic acid (RA) pathway, is a central developmental and tissue homeostasis regulatory signal, strongly dependent on nutritional sources of retinoids and affected by environmental chemicals. This pathway is characterized by multiple proteins or enzymes capable of performing each step and their integration into a self-regulating network. We studied RA network robustness by transient physiological RA signaling disturbances followed by kinetic transcriptomic analysis of the recovery during embryogenesis. The RA metabolic network was identified as the main regulated module to achieve signaling robustness using an unbiased pattern analysis. We describe the network-wide responses to RA signal manipulation and found the feedback autoregulation to be sensitive to the direction of the RA perturbation: RA knockdown exhibited an upper response limit, whereas RA addition had a minimal feedback-activation threshold. Surprisingly, our robustness response analysis suggests that the RA metabolic network regulation exhibits a multi-objective optimization, known as Pareto optimization, characterized by trade-offs between competing functionalities. We observe that efficient robustness to increasing RA is accompanied by worsening robustness to reduced RA levels and vice versa. This direction-dependent trade-off in the network-wide feedback response, results in an uneven robustness capacity of the RA network during early embryogenesis, likely a significant contributor to the manifestation of developmental defects.
Collapse
Affiliation(s)
- Madhur Parihar
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Liat Bendelac-Kapon
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Michal Gur
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Tali Abbou
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Abha Belorkar
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sirisha Achanta
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Keren Kinberg
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| |
Collapse
|
28
|
Qiu Y, Fung L, Schilling TF, Nie Q. Multiple morphogens and rapid elongation promote segmental patterning during development. PLoS Comput Biol 2021; 17:e1009077. [PMID: 34161317 PMCID: PMC8259987 DOI: 10.1371/journal.pcbi.1009077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 07/06/2021] [Accepted: 05/13/2021] [Indexed: 12/21/2022] Open
Abstract
The vertebrate hindbrain is segmented into rhombomeres (r) initially defined by distinct domains of gene expression. Previous studies have shown that noise-induced gene regulation and cell sorting are critical for the sharpening of rhombomere boundaries, which start out rough in the forming neural plate (NP) and sharpen over time. However, the mechanisms controlling simultaneous formation of multiple rhombomeres and accuracy in their sizes are unclear. We have developed a stochastic multiscale cell-based model that explicitly incorporates dynamic morphogenetic changes (i.e. convergent-extension of the NP), multiple morphogens, and gene regulatory networks to investigate the formation of rhombomeres and their corresponding boundaries in the zebrafish hindbrain. During pattern initiation, the short-range signal, fibroblast growth factor (FGF), works together with the longer-range morphogen, retinoic acid (RA), to specify all of these boundaries and maintain accurately sized segments with sharp boundaries. At later stages of patterning, we show a nonlinear change in the shape of rhombomeres with rapid left-right narrowing of the NP followed by slower dynamics. Rapid initial convergence improves boundary sharpness and segment size by regulating cell sorting and cell fate both independently and coordinately. Overall, multiple morphogens and tissue dynamics synergize to regulate the sizes and boundaries of multiple segments during development. In segmental pattern formation, chemical gradients control gene expression in a concentration-dependent manner to specify distinct gene expression domains. Despite the stochasticity inherent to such biological processes, precise and accurate borders form between segmental gene expression domains. Previous work has revealed synergy between gene regulation and cell sorting in sharpening borders that are initially rough. However, it is still poorly understood how size and boundary sharpness of multiple segments are regulated in a tissue that changes dramatically in its morphology as the embryo develops. Here we develop a stochastic multiscale cell-base model to investigate these questions. Two novel strategies synergize to promote accurate segment formation, a combination of long- and short-range morphogens plus rapid tissue convergence, with one responsible for pattern initiation and the other enabling pattern refinement.
Collapse
Affiliation(s)
- Yuchi Qiu
- Department of Mathematics, University of California, Irvine, California, United States of America
| | - Lianna Fung
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, California, United States of America
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, California, United States of America
- * E-mail: (TFS); (QN)
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California, Irvine, California, United States of America
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, California, United States of America
- * E-mail: (TFS); (QN)
| |
Collapse
|
29
|
Stapornwongkul KS, Vincent JP. Generation of extracellular morphogen gradients: the case for diffusion. Nat Rev Genet 2021; 22:393-411. [PMID: 33767424 DOI: 10.1038/s41576-021-00342-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 02/07/2023]
Abstract
Cells within developing tissues rely on morphogens to assess positional information. Passive diffusion is the most parsimonious transport model for long-range morphogen gradient formation but does not, on its own, readily explain scaling, robustness and planar transport. Here, we argue that diffusion is sufficient to ensure robust morphogen gradient formation in a variety of tissues if the interactions between morphogens and their extracellular binders are considered. A current challenge is to assess how the affinity for extracellular binders, as well as other biophysical and cell biological parameters, determines gradient dynamics and shape in a diffusion-based transport system. Technological advances in genome editing, tissue engineering, live imaging and in vivo biophysics are now facilitating measurement of these parameters, paving the way for mathematical modelling and a quantitative understanding of morphogen gradient formation and modulation.
Collapse
|
30
|
Duong TB, Holowiecki A, Waxman JS. Retinoic acid signaling restricts the size of the first heart field within the anterior lateral plate mesoderm. Dev Biol 2021; 473:119-129. [PMID: 33607112 DOI: 10.1016/j.ydbio.2021.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 01/27/2023]
Abstract
Retinoic acid (RA) signaling is required to restrict heart size through limiting the posterior boundary of the vertebrate cardiac progenitor field within the anterior lateral plate mesoderm (ALPM). However, we still do not fully understand how different cardiac progenitor populations that contribute to the developing heart, including earlier-differentiating first heart field (FHF), later-differentiating second heart field (SHF), and neural crest-derived progenitors, are each affected in RA-deficient embryos. Here, we quantified the number of cardiac progenitors and differentiating cardiomyocytes (CMs) in RA-deficient zebrafish embryos. While Nkx2.5+ cells were increased overall in the nascent hearts of RA-deficient embryos, unexpectedly, we found that the major effect within this population was a significant expansion in the number of differentiating FHF CMs. In contrast to the expansion of the FHF, there was a progressive decrease in SHF progenitors at the arterial pole as the heart tube elongated. Temporal differentiation assays and immunostaining in RA-deficient embryos showed that the outflow tracts (OFTs) of the hearts were significantly smaller, containing fewer differentiated SHF-derived ventricular CMs and a complete absence of SHF-derived smooth muscle at later stages. At the venous pole of the heart, pacemaker cells of the sinoatrial node also failed to differentiate in RA-deficient embryos. Interestingly, genetic lineage tracing showed that the number of neural-crest derived CMs was not altered within the enlarged hearts of RA-deficient zebrafish embryos. Altogether, our data show that the enlarged hearts in RA-deficient zebrafish embryos are comprised of an expansion in earlier differentiating FHF-derived CMs coupled with a progressive depletion of the SHF, suggesting RA signaling determines the relative ratios of earlier- and later-differentiation cardiac progenitors within an expanded cardiac progenitor pool.
Collapse
Affiliation(s)
- Tiffany B Duong
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew Holowiecki
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
31
|
Itoh T, Takeuchi M, Sakagami M, Asakawa K, Sumiyama K, Kawakami K, Shimizu T, Hibi M. Gsx2 is required for specification of neurons in the inferior olivary nuclei from Ptf1a-expressing neural progenitors in zebrafish. Development 2020; 147:dev.190603. [PMID: 32928905 DOI: 10.1242/dev.190603] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/03/2020] [Indexed: 11/20/2022]
Abstract
Neurons in the inferior olivary nuclei (IO neurons) send climbing fibers to Purkinje cells to elicit functions of the cerebellum. IO neurons and Purkinje cells are derived from neural progenitors expressing the proneural gene ptf1a In this study, we found that the homeobox gene gsx2 was co-expressed with ptf1a in IO progenitors in zebrafish. Both gsx2 and ptf1a zebrafish mutants showed a strong reduction or loss of IO neurons. The expression of ptf1a was not affected in gsx2 mutants, and vice versa. In IO progenitors, the ptf1a mutation increased apoptosis whereas the gsx2 mutation did not, suggesting that ptf1a and gsx2 are regulated independently of each other and have distinct roles. The fibroblast growth factors (Fgf) 3 and 8a, and retinoic acid signals negatively and positively, respectively, regulated gsx2 expression and thereby the development of IO neurons. mafba and Hox genes are at least partly involved in the Fgf- and retinoic acid-dependent regulation of IO neuronal development. Our results indicate that gsx2 mediates the rostro-caudal positional signals to specify the identity of IO neurons from ptf1a-expressing neural progenitors.
Collapse
Affiliation(s)
- Tsubasa Itoh
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Miki Takeuchi
- Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8601, Japan
| | - Marina Sakagami
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Kazuhide Asakawa
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, 411-8540, Japan
| | - Kenta Sumiyama
- RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka 565-0871, Japan
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics, Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, 411-8540, Japan
| | - Takashi Shimizu
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan.,Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8601, Japan
| | - Masahiko Hibi
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan .,Bioscience and Biotechnology Center, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8601, Japan
| |
Collapse
|
32
|
The Cdx transcription factors and retinoic acid play parallel roles in antero-posterior position of the pectoral fin field during gastrulation. Mech Dev 2020; 164:103644. [PMID: 32911082 DOI: 10.1016/j.mod.2020.103644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/27/2022]
Abstract
The molecular regulators that determine the precise position of the vertebrate limb along the anterio-posterior axis have not been identified. One model suggests that a combination of hox genes in the lateral plate mesoderm (LPM) promotes formation of the limb field, however redundancy among duplicated paralogs has made this model difficult to confirm. In this study, we identify an optimal window during mid-gastrulation stages when transient mis-regulation of retinoic acid signaling or the caudal related transcription factor, Cdx4, both known regulators of hox genes, can alter the position of the pectoral fin field. We show that increased levels of either RA or Cdx4 during mid-gastrulation are sufficient to rostrally shift the position of the pectoral fin field at the expense of surrounding gene expression in the anterior lateral plate mesoderm (aLPM). Alternatively, embryos deficient for both Cdx4 and Cdx1a (Cdx-deficient) form pectoral fins that are shifted towards the posterior and reveal an additional effect on size of the pectoral fin buds. Prior to formation of the pectoral fin buds, the fin field in Cdx-deficient embryos is visibly expanded into the posterior LPM (pLPM) region at the expense of surrounding gene expression. The effects on gene expression immediately post-gastrulation and during somitogenesis support a model where RA and Cdx4 act in parallel to regulate the position of the pectoral fin. Our transient method is a potentially useful model for studying the mechanisms of limb positioning along the AP axis.
Collapse
|
33
|
Fainsod A, Bendelac-Kapon L, Shabtai Y. Fetal Alcohol Spectrum Disorder: Embryogenesis Under Reduced Retinoic Acid Signaling Conditions. Subcell Biochem 2020; 95:197-225. [PMID: 32297301 DOI: 10.1007/978-3-030-42282-0_8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Fetal Alcohol Spectrum Disorder (FASD) is a complex set of developmental malformations, neurobehavioral anomalies and mental disabilities induced by exposing human embryos to alcohol during fetal development. Several experimental models and a series of developmental and biochemical approaches have established a strong link between FASD and reduced retinoic acid (RA) signaling. RA signaling is involved in the regulation of numerous developmental decisions from patterning of the anterior-posterior axis, starting at gastrulation, to the differentiation of specific cell types within developing organs, to adult tissue homeostasis. Being such an important regulatory signal during embryonic development, mutations or environmental perturbations that affect the level, timing or location of the RA signal can induce multiple and severe developmental malformations. The evidence connecting human syndromes to reduced RA signaling is presented here and the resulting phenotypes are compared to FASD. Available data suggest that competition between ethanol clearance and RA biosynthesis is a major etiological component in FASD.
Collapse
Affiliation(s)
- Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel.
| | - Liat Bendelac-Kapon
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel
| | - Yehuda Shabtai
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Ein Kerem, POB 12271, 9112102, Jerusalem, Israel
| |
Collapse
|
34
|
Parker HJ, Krumlauf R. A Hox gene regulatory network for hindbrain segmentation. Curr Top Dev Biol 2020; 139:169-203. [DOI: 10.1016/bs.ctdb.2020.03.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
Cambronero F, Ariza‐McNaughton L, Wiedemann LM, Krumlauf R. Inter‐rhombomeric interactions reveal roles for fibroblast growth factors signaling in segmental regulation of
EphA4
expression. Dev Dyn 2019; 249:354-368. [DOI: 10.1002/dvdy.101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
| | | | - Leanne M. Wiedemann
- Stowers Institute for Medical Research Kansas City Missouri
- Department of Pathology and Laboratory MedicineKansas University Medical Center Kansas City Kansas
| | - Robb Krumlauf
- Stowers Institute for Medical Research Kansas City Missouri
- Division of Developmental NeurobiologyNational Institute for Medical Research London UK
- Department of Anatomy and Cell BiologyKansas University Medical School Kansas City Kansas
| |
Collapse
|
36
|
Abstract
Retinoic acid (RA), a metabolite of retinol (vitamin A), functions as a ligand for nuclear RA receptors (RARs) that regulate development of chordate animals. RA-RARs can activate or repress transcription of key developmental genes. Genetic studies in mouse and zebrafish embryos that are deficient in RA-generating enzymes or RARs have been instrumental in identifying RA functions, revealing that RA signaling regulates development of many organs and tissues, including the body axis, spinal cord, forelimbs, heart, eye and reproductive tract. An understanding of the normal functions of RA signaling during development will guide efforts for use of RA as a therapeutic agent to improve human health. Here, we provide an overview of RA signaling and highlight its key functions during development.
Collapse
Affiliation(s)
- Norbert B Ghyselinck
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Département de Génétique Fonctionnelle et Cancer, Centre National de la Recherche Scientifique (CNRS UMR7104), Institut National de la Santé et de la Recherche Médicale (INSERM U1258), Université de Strasbourg (UNISTRA), 1 rue Laurent Fries, F-67404 Illkirch Cedex, France
| | - Gregg Duester
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
37
|
Williams AL, Bohnsack BL. What's retinoic acid got to do with it? Retinoic acid regulation of the neural crest in craniofacial and ocular development. Genesis 2019; 57:e23308. [PMID: 31157952 DOI: 10.1002/dvg.23308] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/23/2019] [Accepted: 05/05/2019] [Indexed: 12/21/2022]
Abstract
Retinoic acid (RA), the active derivative of vitamin A (retinol), is an essential morphogen signaling molecule and major regulator of embryonic development. The dysregulation of RA levels during embryogenesis has been associated with numerous congenital anomalies, including craniofacial, auditory, and ocular defects. These anomalies result from disruptions in the cranial neural crest, a vertebrate-specific transient population of stem cells that contribute to the formation of diverse cell lineages and embryonic structures during development. In this review, we summarize our current knowledge of the RA-mediated regulation of cranial neural crest induction at the edge of the neural tube and the migration of these cells into the craniofacial region. Further, we discuss the role of RA in the regulation of cranial neural crest cells found within the frontonasal process, periocular mesenchyme, and pharyngeal arches, which eventually form the bones and connective tissues of the head and neck and contribute to structures in the anterior segment of the eye. We then review our understanding of the mechanisms underlying congenital craniofacial and ocular diseases caused by either the genetic or toxic disruption of RA signaling. Finally, we discuss the role of RA in maintaining neural crest-derived structures in postembryonic tissues and the implications of these studies in creating new treatments for degenerative craniofacial and ocular diseases.
Collapse
Affiliation(s)
- Antionette L Williams
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
38
|
Perl E, Waxman JS. Reiterative Mechanisms of Retinoic Acid Signaling during Vertebrate Heart Development. J Dev Biol 2019; 7:jdb7020011. [PMID: 31151214 PMCID: PMC6631158 DOI: 10.3390/jdb7020011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/25/2019] [Accepted: 05/28/2019] [Indexed: 01/07/2023] Open
Abstract
Tightly-regulated levels of retinoic acid (RA) are critical for promoting normal vertebrate development. The extensive history of research on RA has shown that its proper regulation is essential for cardiac progenitor specification and organogenesis. Here, we discuss the roles of RA signaling and its establishment of networks that drive both early and later steps of normal vertebrate heart development. We focus on studies that highlight the drastic effects alternative levels of RA have on early cardiomyocyte (CM) specification and cardiac chamber morphogenesis, consequences of improper RA synthesis and degradation, and known effectors downstream of RA. We conclude with the implications of these findings to our understanding of cardiac regeneration and the etiologies of congenital heart defects.
Collapse
Affiliation(s)
- Eliyahu Perl
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
- The Heart Institute and Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Joshua S Waxman
- The Heart Institute and Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
39
|
Nolte C, De Kumar B, Krumlauf R. Hox genes: Downstream "effectors" of retinoic acid signaling in vertebrate embryogenesis. Genesis 2019; 57:e23306. [PMID: 31111645 DOI: 10.1002/dvg.23306] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022]
Abstract
One of the major regulatory challenges of animal development is to precisely coordinate in space and time the formation, specification, and patterning of cells that underlie elaboration of the basic body plan. How does the vertebrate plan for the nervous and hematopoietic systems, heart, limbs, digestive, and reproductive organs derive from seemingly similar population of cells? These systems are initially established and patterned along the anteroposterior axis (AP) by opposing signaling gradients that lead to the activation of gene regulatory networks involved in axial specification, including the Hox genes. The retinoid signaling pathway is one of the key signaling gradients coupled to the establishment of axial patterning. The nested domains of Hox gene expression, which provide a combinatorial code for axial patterning, arise in part through a differential response to retinoic acid (RA) diffusing from anabolic centers established within the embryo during development. Hence, Hox genes are important direct effectors of retinoid signaling in embryogenesis. This review focuses on describing current knowledge on the complex mechanisms and regulatory processes, which govern the response of Hox genes to RA in several tissue contexts including the nervous system during vertebrate development.
Collapse
Affiliation(s)
- Christof Nolte
- Stowers Institute for Medical Research, Kansas City, Missouri
| | - Bony De Kumar
- Stowers Institute for Medical Research, Kansas City, Missouri
| | - Robb Krumlauf
- Stowers Institute for Medical Research, Kansas City, Missouri.,Department of Anatomy and Cell Biology, Kansas University Medical Center, Kansas City, Kansas
| |
Collapse
|
40
|
Talbot JC, Teets EM, Ratnayake D, Duy PQ, Currie PD, Amacher SL. Muscle precursor cell movements in zebrafish are dynamic and require Six family genes. Development 2019; 146:dev171421. [PMID: 31023879 PMCID: PMC6550023 DOI: 10.1242/dev.171421] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/16/2019] [Indexed: 01/09/2023]
Abstract
Muscle precursors need to be correctly positioned during embryonic development for proper body movement. In zebrafish, a subset of hypaxial muscle precursors from the anterior somites undergo long-range migration, moving away from the trunk in three streams to form muscles in distal locations such as the fin. We mapped long-distance muscle precursor migrations with unprecedented resolution using live imaging. We identified conserved genes necessary for normal precursor motility (six1a, six1b, six4a, six4b and met). These genes are required for movement away from somites and later to partition two muscles within the fin bud. During normal development, the middle muscle precursor stream initially populates the fin bud, then the remainder of this stream contributes to the posterior hypaxial muscle. When we block fin bud development by impairing retinoic acid synthesis or Fgfr function, the entire stream contributes to the posterior hypaxial muscle indicating that muscle precursors are not committed to the fin during migration. Our findings demonstrate a conserved muscle precursor motility pathway, identify dynamic cell movements that generate posterior hypaxial and fin muscles, and demonstrate flexibility in muscle precursor fates.
Collapse
Affiliation(s)
- Jared C Talbot
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA
| | - Emily M Teets
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Dhanushika Ratnayake
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
- EMBL Australia, Monash University, Clayton, VIC, 3800, Australia
| | - Phan Q Duy
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
- EMBL Australia, Monash University, Clayton, VIC, 3800, Australia
| | - Sharon L Amacher
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
- Center for Muscle Health and Neuromuscular Disorders, The Ohio State University and Nationwide Children's Hospital, Columbus, OH 43210, USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
- Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
41
|
Pierdonati E, Unterholzner SJ, Salvi E, Svolacchia N, Bertolotti G, Dello Ioio R, Sabatini S, Di Mambro R. Cytokinin-Dependent Control of GH3 Group II Family Genes in the Arabidopsis Root. PLANTS 2019; 8:plants8040094. [PMID: 30965632 PMCID: PMC6524372 DOI: 10.3390/plants8040094] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/04/2019] [Accepted: 04/06/2019] [Indexed: 01/13/2023]
Abstract
The Arabidopsis root is a dynamic system where the interaction between different plant hormones controls root meristem activity and, thus, organ growth. In the root, a characteristic graded distribution of the hormone auxin provides positional information, coordinating the proliferating and differentiating cell status. The hormone cytokinin shapes this gradient by positioning an auxin minimum in the last meristematic cells. This auxin minimum triggers a cell developmental switch necessary to start the differentiation program, thus, regulating the root meristem size. To position the auxin minimum, cytokinin promotes the expression of the IAA-amido synthase group II gene GH3.17, which conjugates auxin with amino acids, in the most external layer of the root, the lateral root cap tissue. Since additional GH3 genes are expressed in the root, we questioned whether cytokinin to position the auxin minimum also operates via different GH3 genes. Here, we show that cytokinin regulates meristem size by activating the expression of GH3.5 and GH3.6 genes, in addition to GH3.17. Thus, cytokinin activity provides a robust control of auxin activity in the entire organ necessary to regulate root growth.
Collapse
Affiliation(s)
- Emanuela Pierdonati
- Dipartimento di Biologia e Biotecnologie, Laboratory of Functional Genomics and Proteomics of Model Systems, Università di Roma, Sapienza-via dei Sardi, 70⁻00185 Rome, Italy.
| | - Simon Josef Unterholzner
- Dipartimento di Biologia e Biotecnologie, Laboratory of Functional Genomics and Proteomics of Model Systems, Università di Roma, Sapienza-via dei Sardi, 70⁻00185 Rome, Italy.
| | - Elena Salvi
- Dipartimento di Biologia e Biotecnologie, Laboratory of Functional Genomics and Proteomics of Model Systems, Università di Roma, Sapienza-via dei Sardi, 70⁻00185 Rome, Italy.
| | - Noemi Svolacchia
- Dipartimento di Biologia e Biotecnologie, Laboratory of Functional Genomics and Proteomics of Model Systems, Università di Roma, Sapienza-via dei Sardi, 70⁻00185 Rome, Italy.
| | - Gaia Bertolotti
- Dipartimento di Biologia e Biotecnologie, Laboratory of Functional Genomics and Proteomics of Model Systems, Università di Roma, Sapienza-via dei Sardi, 70⁻00185 Rome, Italy.
| | - Raffaele Dello Ioio
- Dipartimento di Biologia e Biotecnologie, Laboratory of Functional Genomics and Proteomics of Model Systems, Università di Roma, Sapienza-via dei Sardi, 70⁻00185 Rome, Italy.
| | - Sabrina Sabatini
- Dipartimento di Biologia e Biotecnologie, Laboratory of Functional Genomics and Proteomics of Model Systems, Università di Roma, Sapienza-via dei Sardi, 70⁻00185 Rome, Italy.
| | - Riccardo Di Mambro
- Department of Biology, University of Pisa-via L. Ghini, 13⁻56126 Pisa, Italy.
| |
Collapse
|
42
|
Frank D, Sela-Donenfeld D. Hindbrain induction and patterning during early vertebrate development. Cell Mol Life Sci 2019; 76:941-960. [PMID: 30519881 PMCID: PMC11105337 DOI: 10.1007/s00018-018-2974-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/28/2022]
Abstract
The hindbrain is a key relay hub of the central nervous system (CNS), linking the bilaterally symmetric half-sides of lower and upper CNS centers via an extensive network of neural pathways. Dedicated neural assemblies within the hindbrain control many physiological processes, including respiration, blood pressure, motor coordination and different sensations. During early development, the hindbrain forms metameric segmented units known as rhombomeres along the antero-posterior (AP) axis of the nervous system. These compartmentalized units are highly conserved during vertebrate evolution and act as the template for adult brainstem structure and function. TALE and HOX homeodomain family transcription factors play a key role in the initial induction of the hindbrain and its specification into rhombomeric cell fate identities along the AP axis. Signaling pathways, such as canonical-Wnt, FGF and retinoic acid, play multiple roles to initially induce the hindbrain and regulate Hox gene-family expression to control rhombomeric identity. Additional transcription factors including Krox20, Kreisler and others act both upstream and downstream to Hox genes, modulating their expression and protein activity. In this review, we will examine the earliest embryonic signaling pathways that induce the hindbrain and subsequent rhombomeric segmentation via Hox and other gene expression. We will examine how these signaling pathways and transcription factors interact to activate downstream targets that organize the segmented AP pattern of the embryonic vertebrate hindbrain.
Collapse
Affiliation(s)
- Dale Frank
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, 31096, Haifa, Israel.
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 76100, Rehovot, Israel.
| |
Collapse
|
43
|
Shukrun N, Shabtai Y, Pillemer G, Fainsod A. Retinoic acid signaling reduction recapitulates the effects of alcohol on embryo size. Genesis 2019; 57:e23284. [PMID: 30672660 DOI: 10.1002/dvg.23284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/13/2019] [Accepted: 01/21/2019] [Indexed: 12/16/2022]
Abstract
Intrauterine growth restriction (IUGR) is commonly observed in human pregnancies and can result in severe clinical outcomes. IUGR is observed in Fetal Alcohol Syndrome (FAS) fetuses as a result of alcohol (ethanol) exposure during pregnancy. To further understand FAS, the severe form of Fetal Alcohol Spectrum Disorder, we performed an extensive quantitative analysis of the effects of ethanol on embryo size utilizing our Xenopus model. Ethanol-treated embryos exhibited size reduction along the anterior-posterior axis. This effect was evident primarily from the hindbrain caudally, while rostral regions appeared refractive to ethanol-induced size changes, also known as asymmetric IUGR. Interestingly, some embryo batches in addition to shortening from the hindbrain caudally also exhibited an alcohol-dependent reduction of the anterior head domain, known as symmetric IUGR. To study the connection between ethanol exposure and reduced retinoic acid levels we treated embryos with the retinaldehyde dehydrogenase inhibitors, DEAB and citral. Inhibition of retinoic acid biosynthesis recapitulated the growth defects induced by ethanol affecting mainly axial elongation from the hindbrain caudally. To study the competition between ethanol clearance and retinoic acid biosynthesis we demonstrated that, co-exposure to alcohol reduces the teratogenic effects of treatment with retinol (vitamin A), the retinoic acid precursor. These results further support the role of retinoic acid in the regulation of axial elongation.
Collapse
Affiliation(s)
- Natalie Shukrun
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yehuda Shabtai
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Graciela Pillemer
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
44
|
Ueda S, Cordeiro IR, Moriyama Y, Nishimori C, Kai KI, Yu R, Nakato R, Shirahige K, Tanaka M. Cux2 refines the forelimb field by controlling expression of Raldh2 and Hox genes. Biol Open 2019; 8:bio.040584. [PMID: 30651234 PMCID: PMC6398465 DOI: 10.1242/bio.040584] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In vertebrates, two pairs of buds that give rise to the fore- and hindlimbs form at discrete positions along the rostral-caudal axis of the body. The mechanism responsible for the positioning of the limb buds is still largely unknown. Here we show a novel function for Cut homeobox transcription factor 2 (Cux2), the ortholog of Drosophila cut, in refining the forelimb field during chick development. Cux2 is expressed in the forelimb field before the emergence of the limb buds. Knocking down the expression of Cux2 using small interfering RNA (siRNA) resulted in a caudal shift of the forelimb bud, whereas misexpression of Cux2 or the constitutively active Cux2-VP16 caused a rostral shift of the forelimb bud or reduction of the forelimb field along the anterior-posterior axis. Further functional analyses revealed that expression of Hoxb genes and retinaldehyde dehydrogenase 2 (Raldh2), which are involved in limb positioning, are directly activated by Cux2 in the lateral plate mesoderm. Our data suggest that Cux2 in the lateral plate mesoderm refines the forelimb field via regulation of Raldh2 and Hoxb genes in chicken embryos. Summary: Cux2 in the lateral plate mesoderm refines the forelimb field via regulation of Raldh2 and Hoxb genes in chicken embryos.
Collapse
Affiliation(s)
- Shogo Ueda
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, 226-8501, Japan
| | - Ingrid Rosenburg Cordeiro
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, 226-8501, Japan
| | - Yuuta Moriyama
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, 226-8501, Japan
| | - Chika Nishimori
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, 226-8501, Japan
| | - Kei-Ichi Kai
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, 226-8501, Japan
| | - Reiko Yu
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, 226-8501, Japan
| | - Ryoichiro Nakato
- Research Center for Epigenetic Disease, Institute of Molecular and Cellular Biosciences, University of Tokyo, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Katsuhiko Shirahige
- Research Center for Epigenetic Disease, Institute of Molecular and Cellular Biosciences, University of Tokyo, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Mikiko Tanaka
- Department of Life Science and Technology, Tokyo Institute of Technology, Midori-ku, Yokohama, 226-8501, Japan
| |
Collapse
|
45
|
Dohn TE, Ravisankar P, Tirera FT, Martin KE, Gafranek JT, Duong TB, VanDyke TL, Touvron M, Barske LA, Crump JG, Waxman JS. Nr2f-dependent allocation of ventricular cardiomyocyte and pharyngeal muscle progenitors. PLoS Genet 2019; 15:e1007962. [PMID: 30721228 PMCID: PMC6377147 DOI: 10.1371/journal.pgen.1007962] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/15/2019] [Accepted: 01/14/2019] [Indexed: 12/28/2022] Open
Abstract
Multiple syndromes share congenital heart and craniofacial muscle defects, indicating there is an intimate relationship between the adjacent cardiac and pharyngeal muscle (PM) progenitor fields. However, mechanisms that direct antagonistic lineage decisions of the cardiac and PM progenitors within the anterior mesoderm of vertebrates are not understood. Here, we identify that retinoic acid (RA) signaling directly promotes the expression of the transcription factor Nr2f1a within the anterior lateral plate mesoderm. Using zebrafish nr2f1a and nr2f2 mutants, we find that Nr2f1a and Nr2f2 have redundant requirements restricting ventricular cardiomyocyte (CM) number and promoting development of the posterior PMs. Cre-mediated genetic lineage tracing in nr2f1a; nr2f2 double mutants reveals that tcf21+ progenitor cells, which can give rise to ventricular CMs and PM, more frequently become ventricular CMs potentially at the expense of posterior PMs in nr2f1a; nr2f2 mutants. Our studies reveal insights into the molecular etiology that may underlie developmental syndromes that share heart, neck and facial defects as well as the phenotypic variability of congenital heart defects associated with NR2F mutations in humans.
Collapse
Affiliation(s)
- Tracy E. Dohn
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Padmapriyadarshini Ravisankar
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Fouley T. Tirera
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Master’s Program in Genetics, Department of Life Sciences, Université Paris Diderot, Paris, France
| | - Kendall E. Martin
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Molecular Genetics and Human Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Jacob T. Gafranek
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Tiffany B. Duong
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Molecular and Developmental Biology Master’s Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Terri L. VanDyke
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Melissa Touvron
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Lindsey A. Barske
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States of America
| | - J. Gage Crump
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States of America
| | - Joshua S. Waxman
- Division of Molecular Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| |
Collapse
|
46
|
Petrelli B, Bendelac L, Hicks GG, Fainsod A. Insights into retinoic acid deficiency and the induction of craniofacial malformations and microcephaly in fetal alcohol spectrum disorder. Genesis 2019; 57:e23278. [DOI: 10.1002/dvg.23278] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Berardino Petrelli
- Regenerative Medicine Program and the Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of Manitoba Winnipeg Manitoba Canada
| | - Liat Bendelac
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel‐CanadaFaculty of Medicine, Hebrew University Jerusalem Israel
| | - Geoffrey G. Hicks
- Regenerative Medicine Program and the Department of Biochemistry & Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health SciencesUniversity of Manitoba Winnipeg Manitoba Canada
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel‐CanadaFaculty of Medicine, Hebrew University Jerusalem Israel
| |
Collapse
|
47
|
Li J, Zhao Y, He L, Huang Y, Yang X, Yu L, Zhao Q, Dong X. Znfl1s are essential for patterning the anterior-posterior axis of zebrafish posterior hindbrain by acting as direct target genes of retinoic acid. Mech Dev 2018; 155:27-33. [PMID: 30472261 DOI: 10.1016/j.mod.2018.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/29/2018] [Accepted: 11/22/2018] [Indexed: 10/27/2022]
Abstract
RA (retinoic acid) signaling is essential for the patterning the hindbrain of vertebrates. Although hundreds of potential RA targets genes are identified, the ones other than hox genes playing roles in patterning anterior-posterior axis of hindbrain by mediating RA signaling remains largely unknown. Previously, we reported that znfl1s play essential roles in the formation of posterior neuroectoderm in zebrafish embryos. Here, we revealed that znfl1s play a critical role in patterning the posterior axis of hindbrain by maintaining the homeostasis of RA signaling in zebrafish embryos. Knocking down znfl1s shortened the length of the posterior hindbrain in a similar way of reducing RA signaling in zebrafish embryos and the defective posterior hindbrain was effectively rescued by elevating RA signaling. By performing mutagenesis assays and chromatin immunoprecipitation assays on the promoter of znfl1s, we demonstrated that znfl1s are direct target genes of RA to mediate RA signaling through a functional DR1 RA response element. Taken together, our results showed that Znfl1s are essential for patterning the anterior-posterior axis development of posterior hindbrain by acting as direct target genes of RA signaling.
Collapse
Affiliation(s)
- Jingyun Li
- Maternal and Child Health Medical Institute, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, China
| | - Yingmin Zhao
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang 214500, China
| | - Luqingqing He
- Model Animal Research Center, Nanjing University, Nanjing 210061, China
| | - Yun Huang
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang 214500, China
| | - Xiaojing Yang
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang 214500, China
| | - Lingling Yu
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang 214500, China
| | - Qingshun Zhao
- Model Animal Research Center, Nanjing University, Nanjing 210061, China.
| | - Xiaohua Dong
- Department of Pediatric, Jingjiang People's Hospital Affiliated to Yangzhou University, Jingjiang 214500, China.
| |
Collapse
|
48
|
Burg L, Palmer N, Kikhi K, Miroshnik ES, Rueckert H, Gaddy E, MacPherson Cunningham C, Mattonet K, Lai SL, Marín-Juez R, Waring RB, Stainier DYR, Balciunas D. Conditional mutagenesis by oligonucleotide-mediated integration of loxP sites in zebrafish. PLoS Genet 2018; 14:e1007754. [PMID: 30427827 PMCID: PMC6261631 DOI: 10.1371/journal.pgen.1007754] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 11/28/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Many eukaryotic genes play essential roles in multiple biological processes in several different tissues. Conditional mutants are needed to analyze genes with such pleiotropic functions. In vertebrates, conditional gene inactivation has only been feasible in the mouse, leaving other model systems to rely on surrogate experimental approaches such as overexpression of dominant negative proteins and antisense-based tools. Here, we have developed a simple and straightforward method to integrate loxP sequences at specific sites in the zebrafish genome using the CRISPR/Cas9 technology and oligonucleotide templates for homology directed repair. We engineered conditional (floxed) mutants of tbx20 and fleer, and demonstrate excision of exons flanked by loxP sites using tamoxifen-inducible CreERT2 recombinase. To demonstrate broad applicability of our method, we also integrated loxP sites into two additional genes, aldh1a2 and tcf21. The ease of this approach will further expand the use of zebrafish to study various aspects of vertebrate biology, especially post-embryonic processes such as regeneration. Some genes are expressed and function in a single tissue, and the effect of their loss on that tissue can be readily determined. Frequently, however, genes that are necessary for the development or maintenance of one tissue are also important for other tissues or cell types. Genes of the latter type are difficult to analyze because of the complications resulting from an organism having multiple defects in different tissues. The solution pioneered by mouse geneticists is to inactivate the gene of interest in only one tissue at a time. This elegant approach requires the ability to make specific edits to the genome, a technology that was not readily available to zebrafish researchers until recently. Using the CRISPR/Cas9 genome editing tool, we have developed a simple, reliable, and efficient method to insert DNA sequences into the zebrafish genome that enable conditional gene inactivation. Our methodology will be useful not only for the study of genes that play important roles in multiple tissues, but also for the genetic analysis of biological processes which occur in adult animals.
Collapse
Affiliation(s)
- Leonard Burg
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Nicholas Palmer
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Khrievono Kikhi
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Evgeniya S. Miroshnik
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Helen Rueckert
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Eleanor Gaddy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Carlee MacPherson Cunningham
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Shih-Lei Lai
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rubén Marín-Juez
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Richard B. Waring
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Darius Balciunas
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
49
|
DeLaurier A. Evolution and development of the fish jaw skeleton. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 8:e337. [PMID: 30378758 DOI: 10.1002/wdev.337] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022]
Abstract
The evolution of the jaw represents a key innovation in driving the diversification of vertebrate body plans and behavior. The pharyngeal apparatus originated as gill bars separated by slits in chordate ancestors to vertebrates. Later, with the acquisition of neural crest, pharyngeal arches gave rise to branchial basket cartilages in jawless vertebrates (agnathans), and later bone and cartilage of the jaw, jaw support, and gills of jawed vertebrates (gnathostomes). Major events in the evolution of jaw structure from agnathans to gnathostomes include axial regionalization of pharyngeal elements and formation of a jaw joint. Hox genes specify the anterior-posterior identity of arches, and edn1, dlx, hand2, Jag1b-Notch2 signaling, and Nr2f factors specify dorsal-ventral identity. The formation of a jaw joint, an important step in the transition from an un-jointed pharynx in agnathans to a hinged jaw in gnathostomes involves interaction between nkx3.2, hand2, and barx1 factors. Major events in jaw patterning between fishes and reptiles include changes to elements of the second pharyngeal arch, including a loss of opercular and branchiostegal ray bones and transformation of the hyomandibula into the stapes. Further changes occurred between reptiles and mammals, including the transformation of the articular and quadrate elements of the jaw joint into the malleus and incus of the middle ear. Fossils of transitional jaw phenotypes can be analyzed from a developmental perspective, and there exists potential to use genetic manipulation techniques in extant taxa to test hypotheses about the evolution of jaw patterning in ancient vertebrates. This article is categorized under: Comparative Development and Evolution > Evolutionary Novelties Early Embryonic Development > Development to the Basic Body Plan Comparative Development and Evolution > Body Plan Evolution.
Collapse
Affiliation(s)
- April DeLaurier
- Department of Biology and Geology, University of South Carolina Aiken, Aiken, South Carolina
| |
Collapse
|
50
|
Berenguer M, Lancman JJ, Cunningham TJ, Dong PDS, Duester G. Mouse but not zebrafish requires retinoic acid for control of neuromesodermal progenitors and body axis extension. Dev Biol 2018; 441:127-131. [PMID: 29964026 DOI: 10.1016/j.ydbio.2018.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/27/2018] [Accepted: 06/27/2018] [Indexed: 12/17/2022]
Abstract
In mouse, retinoic acid (RA) is required for the early phase of body axis extension controlled by a population of neuromesodermal progenitors (NMPs) in the trunk called expanding-NMPs, but not for the later phase of body axis extension controlled by a population of NMPs in the tail called depleting-NMPs. Recent observations suggest that zebrafish utilize depleting-NMPs but not expanding-NMPs for body axis extension. In zebrafish, a role for RA in body axis extension was not supported by previous studies on aldh1a2 (raldh2) mutants lacking RA synthesis. Here, by treating zebrafish embryos with an RA synthesis inhibitor, we also found that body axis extension and somitogenesis was not perturbed, although loss of pectoral fin and cardiac edema were observed consistent with previous studies. The conclusion that zebrafish diverges from mouse in not requiring RA for body axis extension is consistent with zebrafish lacking early expanding-NMPs to generate the trunk. We suggest that RA control of body axis extension was added to higher vertebrates during evolution of expanding-NMPs.
Collapse
Affiliation(s)
- Marie Berenguer
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Joseph J Lancman
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Thomas J Cunningham
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - P Duc Si Dong
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Gregg Duester
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|